1
|
Phadwal K, Haggarty J, Kurian D, Marti JA, Sun J, Houston RD, Betancor MB, MacRae VE, Whitfield PD, Macqueen DJ. Rapamycin induced autophagy enhances lipid breakdown and ameliorates lipotoxicity in Atlantic salmon cells. Biochim Biophys Acta Mol Cell Biol Lipids 2025:159636. [PMID: 40389074 DOI: 10.1016/j.bbalip.2025.159636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 04/22/2025] [Accepted: 05/15/2025] [Indexed: 05/21/2025]
Abstract
Autophagy is a highly conserved cellular recycling process essential for homeostasis in all eukaryotic cells. Lipid accumulation and its regulation by autophagy are key areas of research for understanding metabolic disorders in human and model mammals. However, the role of autophagy in lipid regulation remains poorly characterized in non-model fish species of importance to food production, which could be important for managing health and welfare in aquaculture. Addressing this knowledge gap, we investigate the role of autophagy in lipid regulation using a macrophage-like cell line (SHK-1) from Atlantic salmon (Salmo salar L.), the world's most commercially valuable farmed finfish. Multiple lines of experimental evidence reveal that the autophagic pathway responsible for lipid droplet breakdown is conserved in Atlantic salmon cells. We employed global lipidomics and proteomics analyses on SHK-1 cells subjected to lipid overload, followed by treatment with rapamycin to induce autophagy. This revealed that activating autophagy via rapamycin enhances storage of unsaturated triacylglycerols and suppresses key lipogenic proteins, including fatty acid elongase 6, fatty acid binding protein 2 and acid sphingomyelinase. Moreover, fatty acid elongase 6 and fatty acid binding protein 2 were identified as possible cargo for autophagosomes, suggesting a critical role for autophagy in lipid metabolism in fish. Together, this study establishes a novel model of lipotoxicity and advances understanding of lipid autophagy in fish cells, with significant implications for addressing fish health issues in aquaculture.
Collapse
Affiliation(s)
- Kanchan Phadwal
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK.
| | - Jennifer Haggarty
- Shared Research Facilities, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Dominic Kurian
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - Judit Aguilar Marti
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - Jianxuan Sun
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | | | - Mónica B Betancor
- Institute of Aquaculture, Faculty of Natural Sciences, University of Stirling, Stirling, UK
| | - Vicky E MacRae
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK; School of Life Sciences, Faculty of Science and Engineering, Anglia Ruskin University, Cambridge, UK
| | - Phillip D Whitfield
- Glasgow Polyomics and Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Daniel J Macqueen
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| |
Collapse
|
2
|
Huo Y, Liu X, Lu C, Li T, Yang Z, Xu F, Chen S, Yin K, Wang L. Ceramide mediates cell-to-cell ER stress transmission by modulating membrane fluidity. J Cell Biol 2025; 224:e202405060. [PMID: 40136051 PMCID: PMC11938942 DOI: 10.1083/jcb.202405060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 11/28/2024] [Accepted: 02/13/2025] [Indexed: 03/27/2025] Open
Abstract
Under endoplasmic reticulum (ER) stress (ERS), cells initiate the unfolded protein response (UPR) to maintain ER homeostasis. Recent studies revealed ERS transmission between cells and tissues, by activating the cell-nonautonomous UPR in cells that do not experience ERS directly. Here, we report that ERS triggers a rapid release of ceramide independent of the UPR, but requiring the acid sphingomyelinase activity. Carried by lipoproteins, ceramide is delivered to receiving cells to induce the UPR and regulate cell functions at multiple aspects, including lipid accumulation, cell death, and cytokine production. Mechanistically, extracellular ceramide stimulates ceramide synthesis at the transcription level in receiving cells, leading to ceramide accumulation in the ER so as to reduce membrane fluidity to disrupt ER calcium homeostasis, thus activating the UPR. Sphingomyelin counterbalanced the effect of ceramide. UPR induction is the frontline response to protect cells from ceramide insult. Our study suggests ceramide-mediated ERS transmission as a universal cell-cell communication model regulating a wide range of physiological events.
Collapse
Affiliation(s)
- Yazhen Huo
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Xinlu Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Chen Lu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Tao Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Zaili Yang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Fenfen Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
- Division of Life Sciences and Medicine, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Si Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Kailin Yin
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Likun Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P.R. China
| |
Collapse
|
3
|
Zeng L, Shi W, Chen K, Wang K, Dai Y, Cheng X, Lu S, Gao D, Sun W, Zhang X, Zhang J, Chen J. Indocyanine Green Aggregation-Induced Hypotonic Stress to Remodel Aloe Exosome-like Vesicles for Enhanced Tumor Penetration and Phototherapy. ACS NANO 2025; 19:15425-15443. [PMID: 40243994 DOI: 10.1021/acsnano.4c15440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
As ubiquitous transport nanovesicles in cell biology, plant exosome-like vesicles (PELVs) have enormous potential to deliver drugs safely and effectively. Drug encapsulation and mechanical stability of vesicles are key limitations influencing their delivery efficiency. However, common methods (i.e., ultrasound, electroporation) for drug loading inevitably affect the inherent vesicle characteristics, which influence their stability, leakproof nature, cellular internalization, and tumor penetration. Herein, in order to balance this contradiction, we put forward a strategy to skillfully remodel aloe exosome-like vesicles (AELVs) through indocyanine green (ICG)-induced hypotonic stress during endogenous drug loading. We observe that the rigidity of AELVs is enhanced with the accumulation of long hydrocarbon chain lipids under ICG-induced hypotonic stress. Synchronously, ICG is also loaded into AELVs (ICG/AELVs, IAs), which effectively prevents secondary damage during drug loading. More interestingly, we find that hypotonic stress promotes IA secretion with less intravesicular protein, which is beneficial to enlarge their inner space for more drug loading. The IAs show great storage stability, leakproof, and antidegradation performance. Compared with control AELVs, IAs with higher rigidity are more liable to penetrate into the tumor. IAs further modifying with the AS1411 aptamer (AS1411-IAs, AIAs) exhibit high tumor targeting in vivo. After intravenous administration, the 4T1 tumor is obviously inhibited by AIAs plus NIR irradiation, which effectively improves the survival rate of tumor-bearing mice. Overall, we systematically explore the effects of drug-induced osmotic stress on PELVs during endogenous drug loading and achieve efficient tumor therapy. This work simplifies the process of drug loading in PELVs and enhances their plasticity, which provides a promising perspective for PELV-based drug delivery and clinical application.
Collapse
Affiliation(s)
- Lupeng Zeng
- The School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Innovative Drug Research Institute, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Wanhua Shi
- The School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Innovative Drug Research Institute, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Kewen Chen
- Department of Chemical Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, Fujian, P. R. China
| | - Kun Wang
- The School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Innovative Drug Research Institute, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Yaping Dai
- The School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Innovative Drug Research Institute, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Xin Cheng
- The School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Innovative Drug Research Institute, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Shi Lu
- The School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Innovative Drug Research Institute, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Dandan Gao
- The School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Innovative Drug Research Institute, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Weiming Sun
- The School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Innovative Drug Research Institute, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Xi Zhang
- The School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Innovative Drug Research Institute, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Jing Zhang
- Department of Chemical Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, Fujian, P. R. China
| | - Jinghua Chen
- The School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Innovative Drug Research Institute, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| |
Collapse
|
4
|
Matsushita M, Muri J, Berest I, Li F, Liu H, Corak B, Zamboni N, Buescher J, Othman A, Corrado M, Cupovic J, Werner S, Kovacs W, Kopf M. Peroxisomes are critical for a unique metabolic demand and survival of alveolar macrophages. Cell Rep 2025; 44:115623. [PMID: 40287943 DOI: 10.1016/j.celrep.2025.115623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Tissue-resident macrophages (TRMs) populate throughout various tissues, and their homeostatic metabolism is heavily influenced by these microenvironments. Peroxisomes are organelles that contribute to lipid metabolism. However, the involvement of these organelles in the bioenergetics of TRMs remains undetermined. We conducted a developmental screen of TRMs using a conditional peroxisomal biogenesis factor 5 (Pex5) knockout mouse model that lacks functional peroxisomes in all immune cell subsets. Pulmonary alveolar macrophages (AMs) appeared as the only subset of TRMs that required functional peroxisomes for their development. Pex5 deficiency resulted in reduced AM survival due to increased sensitivity to lipotoxicity, in line with an excess accumulation of ceramides. The absence of peroxisomes had a significant effect on overall mitochondrial fitness and altered their metabolic program, allowing them to engage in glycolysis in addition to oxidative phosphorylation. Our results revealed that AMs have a unique metabolic regulation, where peroxisomes play a central role in their homeostatic development and maintenance.
Collapse
Affiliation(s)
- Mai Matsushita
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Jonathan Muri
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Ivan Berest
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Fengqi Li
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Huan Liu
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Basak Corak
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Nicola Zamboni
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Joerg Buescher
- Metabolomics Facility, Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany
| | - Alaa Othman
- Functional Genomics Center Zurich, Zurich, Switzerland
| | - Mauro Corrado
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Center for Molecular Medicine Cologne and Institute for Genetics, University of Cologne, Cologne, Germany
| | - Jovana Cupovic
- Department of Developmental Immunology, Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany
| | - Sabine Werner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Werner Kovacs
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
5
|
Nepachalovich P, Bonciarelli S, Lombardi Bendoula G, Desantis J, Eleuteri M, Thiele C, Goracci L, Fedorova M. LC-MS and High-Throughput Data Processing Solutions for Lipid Metabolic Tracing Using Bioorthogonal Click Chemistry. Angew Chem Int Ed Engl 2025:e202501884. [PMID: 40270433 DOI: 10.1002/anie.202501884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 04/25/2025]
Abstract
Tracing lipid metabolism in mammalian cells presents a significant technological challenge due to the vast structural diversity of lipids involved in multiple metabolic routes. Bioorthogonal approaches based on click chemistry have revolutionized analytical performance in lipid tracing. When adapted for mass spectrometry (MS), they enable highly specific and sensitive analyses of lipid transformations at the lipidome scale. Here, we advance this approach by integrating liquid chromatography (LC) prior to MS detection and developing a software-assisted workflow for high-throughput data processing. LC separation resolved labeled and unmodified lipids, enabling qualitative and quantitative analysis of both lipidome fractions, as well as isomeric lipid species. Using synthetic standards and endogenously produced alkyne lipids, we characterized LC-MS behavior, including preferential adduct formation and the extent of in-source fragmentation. Specific fragmentation rules, derived from tandem MS experiments for 23 lipid subclasses, were implemented in Lipostar2 software for high-throughput annotation and quantification of labeled lipids. Applying this platform, we traced metabolic pathways of palmitic and oleic acid alkynes, revealing distinct lipid incorporation patterns and metabolic bottlenecks. Altogether, here we provide an integrated analytical and bioinformatics platform for high-throughput tracing of lipid metabolism using LC-MS workflow.
Collapse
Affiliation(s)
- Palina Nepachalovich
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Tatzberg 47-49, Dresden, 01307, Germany
| | | | - Gabriele Lombardi Bendoula
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Tatzberg 47-49, Dresden, 01307, Germany
| | - Jenny Desantis
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, Perugia, 06123, Italy
| | - Michela Eleuteri
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, Perugia, 06123, Italy
| | - Christoph Thiele
- Life & Medical Sciences Institute, University of Bonn, Carl-Troll-Straße 31, Bonn, 53115, Germany
| | - Laura Goracci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, Perugia, 06123, Italy
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Tatzberg 47-49, Dresden, 01307, Germany
| |
Collapse
|
6
|
Xiao M, Zhou N, Tian Z, Sun C. Endogenous Metabolites in Metabolic Diseases: Pathophysiologic Roles and Therapeutic Implications. J Nutr 2025:S0022-3166(25)00227-5. [PMID: 40250565 DOI: 10.1016/j.tjnut.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025] Open
Abstract
Breakthroughs in metabolomics technology have revealed the direct regulatory role of metabolites in physiology and disease. Recent data have highlighted the bioactive metabolites involved in the etiology and prevention and treatment of metabolic diseases such as obesity, nonalcoholic fatty liver disease, type 2 diabetes mellitus, and atherosclerosis. Numerous studies reveal that endogenous metabolites biosynthesized by host organisms or gut microflora regulate metabolic responses and disorders. Lipids, amino acids, and bile acids, as endogenous metabolic modulators, regulate energy metabolism, insulin sensitivity, and immune response through multiple pathways, such as insulin signaling cascade, chemical modifications, and metabolite-macromolecule interactions. Furthermore, the gut microbial metabolites short-chain fatty acids, as signaling regulators have a variety of beneficial impacts in regulating energy metabolic homeostasis. In this review, we will summarize information about the roles of bioactive metabolites in the pathogenesis of many metabolic diseases. Furthermore, we discuss the potential value of metabolites in the promising preventive and therapeutic perspectives of human metabolic diseases.
Collapse
Affiliation(s)
- Mengjie Xiao
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China
| | - Ning Zhou
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China
| | - Zhen Tian
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China.
| | - Changhao Sun
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China.
| |
Collapse
|
7
|
Sehgal R, Jähnert M, Lazaratos M, Speckmann T, Schumacher F, Kleuser B, Ouni M, Jonas W, Schürmann A. Altered liver lipidome markedly overlaps with human plasma lipids at diabetes risk and reveals adipose-liver interaction. J Lipid Res 2025; 66:100767. [PMID: 40044043 PMCID: PMC11997378 DOI: 10.1016/j.jlr.2025.100767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Present study explores the role of liver lipidome in driving T2D-associated metabolic changes. Elevated liver triacylglycerols, reduced PUFAs, and 86 differentially abundant lipid species were identified in diabetes-prone mice. Of these altered lipid species, 82 markedly overlap with human plasma lipids associated with T2D/CVD risk. Pathway enrichment highlighted sphingolipid metabolism, however, only five of all genes involved in the pathway were differentially expressed in the liver. Interestingly, overlap with adipose tissue transcriptome was much higher (57 genes), pointing toward an active adipose-liver interaction. Next, the integration of liver lipidome and transcriptome identified strongly correlated lipid-gene networks highlighting ceramide [Cer(22:0)], dihydroceramide(24:1), and triacylglycerol(58:6) playing a central role in transcriptional regulation. Putative molecular targets of Cer(22:0) were altered (Cyp3a44, Tgf-β1) in primary mouse hepatocytes treated with Cer(22:0). Early alteration of liver lipidome markedly depends on adipose tissue expression pattern and provides substantial evidence linking early liver lipidome alterations and risk of T2D.
Collapse
Affiliation(s)
- Ratika Sehgal
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Department of Internal Medicine 1, University Hospital Ulm, Ulm, Germany
| | - Markus Jähnert
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Michail Lazaratos
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Thilo Speckmann
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | | | - Burkhard Kleuser
- Freie Universität Berlin, Institute of Pharmacy, Berlin, Germany
| | - Meriem Ouni
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Wenke Jonas
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Annette Schürmann
- Department of Experimental Diabetology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.
| |
Collapse
|
8
|
Acosta-Alvear D, Harnoss JM, Walter P, Ashkenazi A. Homeostasis control in health and disease by the unfolded protein response. Nat Rev Mol Cell Biol 2025; 26:193-212. [PMID: 39501044 DOI: 10.1038/s41580-024-00794-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 02/27/2025]
Abstract
Cells rely on the endoplasmic reticulum (ER) to fold and assemble newly synthesized transmembrane and secretory proteins - essential for cellular structure-function and for both intracellular and intercellular communication. To ensure the operative fidelity of the ER, eukaryotic cells leverage the unfolded protein response (UPR) - a stress-sensing and signalling network that maintains homeostasis by rebalancing the biosynthetic capacity of the ER according to need. The metazoan UPR can also redirect signalling from cytoprotective adaptation to programmed cell death if homeostasis restoration fails. As such, the UPR benefits multicellular organisms by preserving optimally functioning cells while removing damaged ones. Nevertheless, dysregulation of the UPR can be harmful. In this Review, we discuss the UPR and its regulatory processes as a paradigm in health and disease. We highlight important recent advances in molecular and mechanistic understanding of the UPR that enable greater precision in designing and developing innovative strategies to harness its potential for therapeutic gain. We underscore the rheostatic character of the UPR, its contextual nature and critical open questions for its further elucidation.
Collapse
Affiliation(s)
| | - Jonathan M Harnoss
- Department of General, Visceral, Thoracic and Transplant Surgery, University Hospital Giessen, Giessen, Germany
| | - Peter Walter
- Altos Labs, Inc., Bay Area Institute of Science, Redwood City, CA, USA.
| | - Avi Ashkenazi
- Research Oncology, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
9
|
Flury A, Aljayousi L, Park HJ, Khakpour M, Mechler J, Aziz S, McGrath JD, Deme P, Sandberg C, González Ibáñez F, Braniff O, Ngo T, Smith S, Velez M, Ramirez DM, Avnon-Klein D, Murray JW, Liu J, Parent M, Mingote S, Haughey NJ, Werneburg S, Tremblay MÈ, Ayata P. A neurodegenerative cellular stress response linked to dark microglia and toxic lipid secretion. Neuron 2025; 113:554-571.e14. [PMID: 39719704 DOI: 10.1016/j.neuron.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 10/22/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024]
Abstract
The brain's primary immune cells, microglia, are a leading causal cell type in Alzheimer's disease (AD). Yet, the mechanisms by which microglia can drive neurodegeneration remain unresolved. Here, we discover that a conserved stress signaling pathway, the integrated stress response (ISR), characterizes a microglia subset with neurodegenerative outcomes. Autonomous activation of ISR in microglia is sufficient to induce early features of the ultrastructurally distinct "dark microglia" linked to pathological synapse loss. In AD models, microglial ISR activation exacerbates neurodegenerative pathologies and synapse loss while its inhibition ameliorates them. Mechanistically, we present evidence that ISR activation promotes the secretion of toxic lipids by microglia, impairing neuron homeostasis and survival in vitro. Accordingly, pharmacological inhibition of ISR or lipid synthesis mitigates synapse loss in AD models. Our results demonstrate that microglial ISR activation represents a neurodegenerative phenotype, which may be sustained, at least in part, by the secretion of toxic lipids.
Collapse
Affiliation(s)
- Anna Flury
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Leen Aljayousi
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Hye-Jin Park
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | | | - Jack Mechler
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biochemistry, CUNY Graduate Center, New York, NY 10016, USA
| | - Siaresh Aziz
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Jackson D McGrath
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Michigan Medicine, Ann Arbor, MI 48105, USA
| | - Pragney Deme
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Colby Sandberg
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C4, Canada
| | | | - Olivia Braniff
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C4, Canada
| | - Thi Ngo
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Simira Smith
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Matthew Velez
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Denice Moran Ramirez
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Dvir Avnon-Klein
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - John W Murray
- Columbia Center for Human Development, Center for Stem Cell Therapies, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jia Liu
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Martin Parent
- CERVO Brain Research Center, Québec City, QC G1E 1T2, Canada
| | - Susana Mingote
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Norman J Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sebastian Werneburg
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Michigan Medicine, Ann Arbor, MI 48105, USA; Michigan Neuroscience Institute, Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C4, Canada; Department of Molecular Medicine, Université Laval, Québec City, QC G1V 0A6, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, QC H3A 2B4, Canada; Canada Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 2A1, Canada; Centre for Advanced Materials and Related Technology and Institute on Aging and Lifelong Health, University of Victoria, Victoria, BC V8N 5M8, Canada
| | - Pinar Ayata
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA; Graduate Program in Biochemistry, CUNY Graduate Center, New York, NY 10016, USA.
| |
Collapse
|
10
|
Wang Y, Yung P, Lu G, Liu Y, Ding C, Mao C, Li ZA, Tuan RS. Musculoskeletal Organs-on-Chips: An Emerging Platform for Studying the Nanotechnology-Biology Interface. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2401334. [PMID: 38491868 PMCID: PMC11733728 DOI: 10.1002/adma.202401334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Nanotechnology-based approaches are promising for the treatment of musculoskeletal (MSK) disorders, which present significant clinical burdens and challenges, but their clinical translation requires a deep understanding of the complex interplay between nanotechnology and MSK biology. Organ-on-a-chip (OoC) systems have emerged as an innovative and versatile microphysiological platform to replicate the dynamics of tissue microenvironment for studying nanotechnology-biology interactions. This review first covers recent advances and applications of MSK OoCs and their ability to mimic the biophysical and biochemical stimuli encountered by MSK tissues. Next, by integrating nanotechnology into MSK OoCs, cellular responses and tissue behaviors may be investigated by precisely controlling and manipulating the nanoscale environment. Analysis of MSK disease mechanisms, particularly bone, joint, and muscle tissue degeneration, and drug screening and development of personalized medicine may be greatly facilitated using MSK OoCs. Finally, future challenges and directions are outlined for the field, including advanced sensing technologies, integration of immune-active components, and enhancement of biomimetic functionality. By highlighting the emerging applications of MSK OoCs, this review aims to advance the understanding of the intricate nanotechnology-MSK biology interface and its significance in MSK disease management, and the development of innovative and personalized therapeutic and interventional strategies.
Collapse
Affiliation(s)
- Yuwen Wang
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Patrick Yung
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Gang Lu
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- School of Biomedical SciencesThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Yuwei Liu
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- The First Affiliated Hospital of Shenzhen UniversityShenzhen Second People's HospitalShenzhenGuangdong518037P. R. China
| | - Changhai Ding
- Clinical Research CentreZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510260China
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmania7000Australia
| | - Chuanbin Mao
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| | - Zhong Alan Li
- Department of Biomedical EngineeringThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- School of Biomedical SciencesThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Key Laboratory of Regenerative MedicineMinistry of EducationSchool of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077P. R. China
- Shenzhen Research InstituteThe Chinese University of Hong KongShenzhen518172P. R. China
| | - Rocky S. Tuan
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkNTHong Kong SAR999077P. R. China
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
- School of Biomedical SciencesThe Chinese University of Hong KongNTHong Kong SAR999077P. R. China
| |
Collapse
|
11
|
Sowton AP, Holzner LMW, Krause FN, Baxter R, Mocciaro G, Krzyzanska DK, Minnion M, O'Brien KA, Harrop MC, Darwin PM, Thackray BD, Vacca M, Feelisch M, Griffin JL, Murray AJ. Chronic inorganic nitrate supplementation does not improve metabolic health and worsens disease progression in mice with diet-induced obesity. Am J Physiol Endocrinol Metab 2025; 328:E69-E91. [PMID: 39653040 DOI: 10.1152/ajpendo.00256.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/16/2024] [Accepted: 11/07/2024] [Indexed: 01/11/2025]
Abstract
Inorganic nitrate (NO3-) has been proposed to be of therapeutic use as a dietary supplement in obesity and related conditions including the metabolic syndrome (MetS), type II diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD). Administration of NO3- to endothelial nitric oxide synthase-deficient mice reversed aspects of MetS; however, the impact of NO3- supplementation in diet-induced obesity is not well understood. Here we investigated the whole body metabolic phenotype and cardiac and hepatic metabolism in mice fed a high-fat, high-sucrose (HFHS) diet for up to 12 mo of age, supplemented with 1 mM NaNO3 (or NaCl) in their drinking water. HFHS feeding was associated with a progressive obesogenic and diabetogenic phenotype, which was not ameliorated by NO3-. Furthermore, HFHS-fed mice supplemented with NO3- showed elevated levels of cardiac fibrosis and accelerated progression of MASLD including development of hepatocellular carcinoma in comparison with NaCl-supplemented mice. NO3- did not enhance mitochondrial β-oxidation capacity in any tissue assayed and did not suppress hepatic lipid accumulation, suggesting it does not prevent lipotoxicity. We conclude that NO3- is ineffective in preventing the metabolic consequences of an obesogenic diet and may instead be detrimental to metabolic health against the background of HFHS feeding. This is the first report of an unfavorable effect of long-term nitrate supplementation in the context of the metabolic challenges of overfeeding, warranting urgent further investigation into the mechanism of this interaction.NEW & NOTEWORTHY Inorganic nitrate has been suggested to be of therapeutic benefit in obesity-related conditions, as it increases nitric oxide bioavailability, enhances mitochondrial β-oxidation, and reverses metabolic syndrome in eNOS-/- mice. However, we here show that over 12 months nitrate was ineffective in preventing metabolic consequences in high fat, high sucrose-fed mice and worsened aspects of metabolic health, impairing cholesterol handling, increasing cardiac fibrosis, and exacerbating steatotic liver disease progression, with acceleration to hepatocellular carcinoma.
Collapse
Affiliation(s)
- Alice P Sowton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Lorenz M W Holzner
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Fynn N Krause
- Department of Biochemistry and Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - Ruby Baxter
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Gabriele Mocciaro
- Department of Biochemistry and Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - Dominika K Krzyzanska
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Magdalena Minnion
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Katie A O'Brien
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Matthew C Harrop
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Paula M Darwin
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Benjamin D Thackray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Michele Vacca
- Department of Biochemistry and Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Martin Feelisch
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Julian L Griffin
- Department of Biochemistry and Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
- The Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
12
|
Feng L, Li B, Yong SS, Wen X, Tian Z. The emerging role of exercise in Alzheimer's disease: Focus on mitochondrial function. Ageing Res Rev 2024; 101:102486. [PMID: 39243893 DOI: 10.1016/j.arr.2024.102486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by memory impairment and cognitive dysfunction, which eventually leads to the disability and mortality of older adults. Although the precise mechanisms by which age promotes the development of AD remains poorly understood, mitochondrial dysfunction plays a central role in the development of AD. Currently, there is no effective treatment for this debilitating disease. It is well accepted that exercise exerts neuroprotective effects by ameliorating mitochondrial dysfunction in the neurons of AD, which involves multiple mechanisms, including mitochondrial dynamics, biogenesis, mitophagy, transport, and signal transduction. In addition, exercise promotes mitochondria communication with other organelles in AD neurons, which should receive more attentions in the future.
Collapse
Affiliation(s)
- Lili Feng
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China.
| | - Bowen Li
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China
| | - Su Sean Yong
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China
| | - Xu Wen
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China.
| | - Zhenjun Tian
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
13
|
Brito ML, Coutinho-Wolino KS, Almeida PP, Trigueira PDC, Alves APDP, Magliano DC, Stockler-Pinto MB. Unstressing the Reticulum: Nutritional Strategies for Modulating Endoplasmic Reticulum Stress in Obesity. Mol Nutr Food Res 2024; 68:e2400361. [PMID: 39363792 DOI: 10.1002/mnfr.202400361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/03/2024] [Indexed: 10/05/2024]
Abstract
The progression of obesity involves several molecular mechanisms that are closely associated with the pathophysiological response of the disease. Endoplasmic reticulum (ER) stress is one such factor. Lipotoxicity disrupts endoplasmic reticulum homeostasis in the context of obesity. Furthermore, it induces ER stress by activating several signaling pathways via inflammatory responses and oxidative stress. ER performs crucial functions in protein synthesis and lipid metabolism; thus, triggers such as lipotoxicity can promote the accumulation of misfolded proteins in the organelle. The accumulation of these proteins can lead to metabolic disorders and chronic inflammation, resulting in cell death. Thus, alternatives, such as flavonoids, amino acids, and polyphenols that are associated with antioxidant and anti-inflammatory responses have been proposed to attenuate this response by modulating ER stress via the administration of nutrients and bioactive compounds. Decreasing inflammation and oxidative stress can reduce the expression of several ER stress markers and improve clinical outcomes through the management of obesity, including the control of body weight, visceral fat, and lipid accumulation. This review explores the metabolic changes resulting from ER stress and discusses the role of nutritional interventions in modulating the ER stress pathway in obesity.
Collapse
Affiliation(s)
- Michele Lima Brito
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
| | - Karen Salve Coutinho-Wolino
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
| | - Patricia Pereira Almeida
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
| | | | - Ana Paula de Paula Alves
- Endocrinology Post Graduate Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 24210-201, Brazil
| | - D'Angelo Carlo Magliano
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Endocrinology Post Graduate Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 24210-201, Brazil
- Morphology Department, Biomedical Institute, Fluminense Federal University (UFF), Niterói, RJ, 24020-150, Brazil
| | - Milena Barcza Stockler-Pinto
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Nutrition Sciences Postgraduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24020-140, Brazil
| |
Collapse
|
14
|
Faccioli LA, Sun Y, Animasahun O, Motomura T, Liu Z, Kurihara T, Hu Z, Yang B, Cetin Z, Baratta AM, Shankaran A, Nenwani M, Altay LN, Huang L, Meurs N, Franks J, Stolz D, Gavlock DC, Miedel MT, Ostrowska A, Florentino RM, Fox IJ, Nagrath D, Soto-Gutierrez A. Human-induced pluripotent stem cell-based hepatic modeling of lipid metabolism-associated TM6SF2-E167K variant. Hepatology 2024:01515467-990000000-01008. [PMID: 39190693 PMCID: PMC11865362 DOI: 10.1097/hep.0000000000001065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/26/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND AND AIMS TM6SF2 rs58542926 (E167K) is related to an increased prevalence of metabolic dysfunction-associated steatotic liver disease. Conflicting mouse study results highlight the need for a human model to understand this mutation's impact. This study aims to create and characterize a reliable human in vitro model to mimic the effects of the TM6SF2-E167K mutation for future studies. APPROACH AND RESULTS We used gene editing on human-induced pluripotent stem cells from a healthy individual to create cells with the TM6SF2-E167K mutation. After hepatocyte-directed differentiation, we observed decreased TM6SF2 protein expression, increased intracellular lipid droplets, and total cholesterol, in addition to reduced VLDL secretion. Transcriptomics revealed the upregulation of genes involved in lipid, fatty acid, and cholesterol transport, flux, and oxidation. Global lipidomics showed increased lipid classes associated with endoplasmic reticulum (ER) stress, mitochondrial dysfunction, apoptosis, and lipid metabolism. In addition, the TM6SF2-E167K mutation conferred a proinflammatory phenotype with signs of mitochondria and ER stress. Importantly, by facilitating protein folding within the ER of hepatocytes carrying TM6SF2-E167K mutation, VLDL secretion and ER stress markers improved. CONCLUSIONS Our findings indicate that induced hepatocytes generated from human-induced pluripotent stem cells carrying the TM6SF2-E167K recapitulate the effects observed in human hepatocytes from individuals with the TM6SF2 mutation. This study characterizes an in vitro model that can be used as a platform to identify potential clinical targets and highlights the therapeutic potential of targeting protein misfolding to alleviate ER stress and mitigate the detrimental effects of the TM6SF2-E167K mutation on hepatic lipid metabolism.
Collapse
Affiliation(s)
- Lanuza A.P. Faccioli
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yiyue Sun
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- School of Medicine, Tsinghua University, Beijing, PRC
| | - Olamide Animasahun
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Takashi Motomura
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Zhenghao Liu
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Takeshi Kurihara
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Zhiping Hu
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Bo Yang
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Zeliha Cetin
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Annalisa M. Baratta
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ajay Shankaran
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Minal Nenwani
- Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Leyla Nurcihan Altay
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Linqi Huang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Noah Meurs
- Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Jonathan Franks
- Department of Cell Biology and Physiology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Donna Stolz
- Department of Cell Biology and Physiology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dillon C. Gavlock
- Drug Discovery Institute, Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mark T. Miedel
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Drug Discovery Institute, Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alina Ostrowska
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rodrigo M. Florentino
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ira J. Fox
- Department of Surgery, Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Deepak Nagrath
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Alejandro Soto-Gutierrez
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pathology, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
15
|
Liepinsh E, Zvejniece L, Clemensson L, Ozola M, Vavers E, Cirule H, Korzh S, Skuja S, Groma V, Briviba M, Grinberga S, Liu W, Olszewski P, Gentreau M, Fredriksson R, Dambrova M, Schiöth HB. Hydroxymethylglutaryl-CoA reductase activity is essential for mitochondrial β-oxidation of fatty acids to prevent lethal accumulation of long-chain acylcarnitines in the mouse liver. Br J Pharmacol 2024; 181:2750-2773. [PMID: 38641905 DOI: 10.1111/bph.16363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/16/2024] [Accepted: 01/30/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND AND PURPOSE Statins are competitive inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase (HMGCR), and exert adverse effects on mitochondrial function, although the mechanisms underlying these effects remain unclear. We used a tamoxifen-induced Hmgcr-knockout (KO) mouse model, a multi-omics approach and mitochondrial function assessments to investigate whether decreased HMGCR activity impacts key liver energy metabolism pathways. EXPERIMENTAL APPROACH We established a new mouse strain using the Cre/loxP system, which enabled whole-body deletion of Hmgcr expression. These mice were crossed with Rosa26Cre mice and treated with tamoxifen to delete Hmgcr in all cells. We performed transcriptomic and metabolomic analyses and thus evaluated time-dependent changes in metabolic functions to identify the pathways leading to cell death in Hmgcr-KO mice. KEY RESULTS Lack of Hmgcr expression resulted in lethality, due to acute liver damage caused by rapid disruption of mitochondrial fatty acid β-oxidation and very high accumulation of long-chain (LC) acylcarnitines in both male and female mice. Gene expression and KO-related phenotype changes were not observed in other tissues. The progression to liver failure was driven by diminished peroxisome formation, which resulted in impaired mitochondrial and peroxisomal fatty acid metabolism, enhanced glucose utilization and whole-body hypoglycaemia. CONCLUSION AND IMPLICATIONS Our findings suggest that HMGCR is crucial for maintaining energy metabolism balance, and its activity is necessary for functional mitochondrial β-oxidation. Moreover, statin-induced adverse reactions might be rescued by the prevention of LC acylcarnitine accumulation.
Collapse
Affiliation(s)
- Edgars Liepinsh
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Riga Stradins University, Riga, Latvia
| | | | | | - Melita Ozola
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Riga Stradins University, Riga, Latvia
| | - Edijs Vavers
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Helena Cirule
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | | | | | | | - Monta Briviba
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | - Wen Liu
- Uppsala University, Uppsala, Sweden
| | | | | | | | - Maija Dambrova
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Riga Stradins University, Riga, Latvia
| | | |
Collapse
|
16
|
Doll CL, Snider AJ. The diverse roles of sphingolipids in inflammatory bowel disease. FASEB J 2024; 38:e23777. [PMID: 38934445 PMCID: PMC467036 DOI: 10.1096/fj.202400830r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024]
Abstract
The incidence of inflammatory bowel disease (IBD) has increased over the last 20 years. A variety of causes, both physiological and environmental, contribute to the initiation and progression of IBD, making disease management challenging. Current treatment options target various aspects of the immune response to dampen intestinal inflammation; however, their effectiveness at retaining remission, their side effects, and loss of response from patients over time warrant further investigation. Finding a common thread within the multitude causes of IBD is critical in developing robust treatment options. Sphingolipids are evolutionary conserved bioactive lipids universally generated in all cell types. This diverse lipid family is involved in a variety of fundamental, yet sometimes opposing, processes such as proliferation and apoptosis. Implicated as regulators in intestinal diseases, sphingolipids are a potential cornerstone in understanding IBD. Herein we will describe the role of host- and microbial-derived sphingolipids as they relate to the many factors of intestinal health and IBD.
Collapse
Affiliation(s)
- Chelsea L. Doll
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ 85721, USA
| | - Ashley J. Snider
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ 85721, USA
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
17
|
Ding S, Li G, Fu T, Zhang T, Lu X, Li N, Geng Q. Ceramides and mitochondrial homeostasis. Cell Signal 2024; 117:111099. [PMID: 38360249 DOI: 10.1016/j.cellsig.2024.111099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 02/17/2024]
Abstract
Lipotoxicity arises from the accumulation of lipid intermediates in non-adipose tissue, precipitating cellular dysfunction and death. Ceramide, a toxic byproduct of excessive free fatty acids, has been widely recognized as a primary contributor to lipotoxicity, mediating various cellular processes such as apoptosis, differentiation, senescence, migration, and adhesion. As the hub of lipid metabolism, the excessive accumulation of ceramides inevitably imposes stress on the mitochondria, leading to the disruption of mitochondrial homeostasis, which is typified by adequate ATP production, regulated oxidative stress, an optimal quantity of mitochondria, and controlled mitochondrial quality. Consequently, this review aims to collate current knowledge and facts regarding the involvement of ceramides in mitochondrial energy metabolism and quality control, thereby providing insights for future research.
Collapse
Affiliation(s)
- Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Guorui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tinglv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tianyu Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiao Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
18
|
Wajapeyee N, Beamon TC, Gupta R. Roles and therapeutic targeting of ceramide metabolism in cancer. Mol Metab 2024; 83:101936. [PMID: 38599378 PMCID: PMC11031839 DOI: 10.1016/j.molmet.2024.101936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Ceramides are sphingolipids that act as signaling molecules involved in regulating cellular processes including apoptosis, proliferation, and metabolism. Deregulation of ceramide metabolism contributes to cancer development and progression. Therefore, regulation of ceramide levels in cancer cells is being explored as a new approach for cancer therapy. SCOPE OF THE REVIEW This review discusses the multiple roles of ceramides in cancer cells and strategies to modulate ceramide levels for cancer therapy. Ceramides attenuate cell survival signaling and metabolic pathways, while activating apoptotic mechanisms, making them tumor-suppressive. Approaches to increase ceramide levels in cancer cells include using synthetic analogs, inhibiting ceramide degradation, and activating ceramide synthesis. We also highlight combination therapies such as use of ceramide modulators with chemotherapies, immunotherapies, apoptosis inducers, and anti-angiogenics, which offer synergistic antitumor effects. Additionally, we also describe ongoing clinical trials evaluating ceramide nanoliposomes and analogs. Finally, we discuss the challenges of these therapeutic approaches including the complexity of ceramide metabolism, targeted delivery, cancer heterogeneity, resistance mechanisms, and long-term safety. MAJOR CONCLUSIONS Ceramide-based therapy is a potentially promising approach for cancer therapy. However, overcoming hurdles in pharmacokinetics, specificity, and resistance is needed to optimize its efficacy and safety. This requires comprehensive preclinical/clinical studies into ceramide signaling, formulations, and combination therapies. Ceramide modulation offers opportunities for developing novel cancer treatments, but a deeper understanding of ceramide biology is vital to advance its clinical applications.
Collapse
Affiliation(s)
- Narendra Wajapeyee
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35233, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| | - Teresa Chiyanne Beamon
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Romi Gupta
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35233, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|
19
|
Liu Y, Sun Z, Sun Q, Wang L, Wang C, Li Y, Ma C, Shi W, Zhang G, Dong Y, Zhang X, Cong B. The effects of restraint stress on ceramide metabolism disorders in the rat liver: the role of CerS6 in hepatocyte injury. Lipids Health Dis 2024; 23:68. [PMID: 38431645 PMCID: PMC10908211 DOI: 10.1186/s12944-024-02019-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/15/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Stress is implicated in various pathological conditions leading to liver injury. Existing evidence suggests that excessive stress can induce mitochondrial damage in hepatocytes, yet the underlying mechanism remains unclear. Ceramide synthase 6 (CerS6)-derived C16:0 ceramide is recognised as a lipotoxic substance capable of causing mitochondrial damage. However, the role of CerS6 in stress has received insufficient attention. This study aimed to explore the involvement of CerS6 in stress-induced hepatic damage and its associated mechanisms. METHODS The rat restraint stress model and a corticosterone (CORT)-induced hepatocyte stress model were employed for in vivo and in vitro experimental analyses, respectively. Changes in mitochondrial damage and ceramide metabolism in hepatocytes induced by stress were evaluated. The impact of CORT on mitochondrial damage and ceramide metabolism in hepatocytes was assessed following CerS6 knockdown. Mitochondria were isolated using a commercial kit, and ceramides in liver tissue and hepatocytes were detected by LC-MS/MS. RESULTS In comparison to the control group, rats subjected to one week of restraint exhibited elevated serum CORT levels. The liver displayed significant signs of mitochondrial damage, accompanied by increased CerS6 and mitochondrial C16:0 ceramide, along with activation of the AMPK/p38 MAPK pathway. In vitro studies demonstrated that CORT treatment of hepatocytes resulted in mitochondrial damage, concomitant with elevated CerS6 and mitochondrial C16:0 ceramide. Furthermore, CORT induced sequential phosphorylation of AMPK and p38 MAPK proteins, and inhibition of the p38 MAPK pathway using SB203580 mitigated the CORT-induced elevation in CerS6 protein. Knocking down CerS6 in hepatocytes inhibited both the increase in C16:0 ceramide and the release of mitochondrial cytochrome c induced by CORT. CONCLUSIONS CerS6-associated C16:0 ceramide plays a mediating role in stress-induced mitochondrial damage in hepatocytes. The molecular mechanism is linked to CORT-induced activation of the AMPK/p38 MAPK pathway, leading to upregulated CerS6.
Collapse
Affiliation(s)
- Yichang Liu
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
- Department of Forensic Medicine, College of Medicine, Nantong University, Nantong, 226000, China
| | - Zhaoling Sun
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Qiuli Sun
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Li Wang
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Chuan Wang
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Yingmin Li
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Chunling Ma
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Weibo Shi
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Guozhong Zhang
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
- Hebei Province Laboratory of Experimental Animal, Shijiazhuang, 050017, China
| | - Yiming Dong
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Xiaojing Zhang
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China.
| | - Bin Cong
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China.
- Hainan Tropical Forensic Medicine Academician Workstation, Haikou, 571199, China.
| |
Collapse
|
20
|
Mu J, Lam SM, Shui G. Emerging roles and therapeutic potentials of sphingolipids in pathophysiology: emphasis on fatty acyl heterogeneity. J Genet Genomics 2024; 51:268-278. [PMID: 37364711 DOI: 10.1016/j.jgg.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/29/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023]
Abstract
Sphingolipids not only exert structural roles in cellular membranes, but also act as signaling molecules in various physiological and pathological processes. A myriad of studies have shown that abnormal levels of sphingolipids and their metabolic enzymes are associated with a variety of human diseases. Moreover, blood sphingolipids can also be used as biomarkers for disease diagnosis. This review summarizes the biosynthesis, metabolism, and pathological roles of sphingolipids, with emphasis on the biosynthesis of ceramide, the precursor for the biosynthesis of complex sphingolipids with different fatty acyl chains. The possibility of using sphingolipids for disease prediction, diagnosis, and treatment is also discussed. Targeting endogenous ceramides and complex sphingolipids along with their specific fatty acyl chain to promote future drug development will also be discussed.
Collapse
Affiliation(s)
- Jinming Mu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Lipidall Technologies Company Limited, Changzhou, Jiangsu 213000, China.
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
21
|
Raber J, Holden S, Kessler K, Glaeser B, McQuesten C, Chaudhari M, Stenzel F, Lenarczyk M, Leonard SW, Morré J, Choi J, Kronenberg A, Borg A, Kwok A, Stevens JF, Olsen C, Willey JS, Bobe G, Minnier J, Baker JE. Effects of photon irradiation in the presence and absence of hindlimb unloading on the behavioral performance and metabolic pathways in the plasma of Fischer rats. Front Physiol 2024; 14:1316186. [PMID: 38260101 PMCID: PMC10800373 DOI: 10.3389/fphys.2023.1316186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction: The space environment astronauts experience during space missions consists of multiple environmental challenges, including microgravity. In this study, we assessed the behavioral and cognitive performances of male Fisher rats 2 months after sham irradiation or total body irradiation with photons in the absence or presence of simulated microgravity. We analyzed the plasma collected 9 months after sham irradiation or total body irradiation for distinct alterations in metabolic pathways and to determine whether changes to metabolic measures were associated with specific behavioral and cognitive measures. Methods: A total of 344 male Fischer rats were irradiated with photons (6 MeV; 3, 8, or 10 Gy) in the absence or presence of simulated weightlessness achieved using hindlimb unloading (HU). To identify potential plasma biomarkers of photon radiation exposure or the HU condition for behavioral or cognitive performance, we performed regression analyses. Results: The behavioral effects of HU on activity levels in an open field, measures of anxiety in an elevated plus maze, and anhedonia in the M&M consumption test were more pronounced than those of photon irradiation. Phenylalanine, tyrosine, and tryptophan metabolism, and phenylalanine metabolism and biosynthesis showed very strong pathway changes, following photon irradiation and HU in animals irradiated with 3 Gy. Here, 29 out of 101 plasma metabolites were associated with 1 out of 13 behavioral measures. In the absence of HU, 22 metabolites were related to behavioral and cognitive measures. In HU animals that were sham-irradiated or irradiated with 8 Gy, one metabolite was related to behavioral and cognitive measures. In HU animals irradiated with 3 Gy, six metabolites were related to behavioral and cognitive measures. Discussion: These data suggest that it will be possible to develop stable plasma biomarkers of behavioral and cognitive performance, following environmental challenges like HU and radiation exposure.
Collapse
Affiliation(s)
- Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- Departments of Neurology, and Radiation Medicine, Division of Neuroscience ONPRC, Oregon Health & Science University, Portland, OR, United States
- College of Pharmacy, Oregon State University, Corvallis, OR, United States
| | - Sarah Holden
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Kat Kessler
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Breanna Glaeser
- Neuroscience Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Chloe McQuesten
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Mitali Chaudhari
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Fiona Stenzel
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Marek Lenarczyk
- Radiation Biosciences Laboratory, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Scott Willem Leonard
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Jeffrey Morré
- Mass Spectrometry Core, Oregon State University, Corvallis, OR, United States
| | - Jaewoo Choi
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Amy Kronenberg
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Alexander Borg
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Andy Kwok
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Jan Frederik Stevens
- College of Pharmacy, Oregon State University, Corvallis, OR, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Christopher Olsen
- Neuroscience Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jeffrey S. Willey
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Gerd Bobe
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
- Department of Animal Sciences, Oregon State University, Corvallis, OR, United States
| | - Jessica Minnier
- Oregon Health & Science University-Portland State University School of Public Health, Knight Cancer Institute Biostatistics Shared Resource, The Knight Cardiovascular Institute, OR Health & Science University, Portland, OR, United States
| | - John E. Baker
- Neuroscience Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
22
|
Ma X, Tan M, Li L, Zhong Z, Li P, Liang J, Song Q. Ni-catalysed assembly of axially chiral alkenes from alkynyl tetracoordinate borons via 1,3-metallate shift. Nat Chem 2024; 16:42-53. [PMID: 38182763 DOI: 10.1038/s41557-023-01396-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 11/13/2023] [Indexed: 01/07/2024]
Abstract
Asymmetric synthesis based on a metallate shift of tetracoordinate borons is an intriguing and challenging topic. Despite the construction of central chirality from tetracoordinate boron species via a 1,2-metallate shift, catalytic asymmetric synthesis of axially chiral compounds from such boron 'ate' complexes is an ongoing challenge. Axially chiral alkenes have received great attention due to their unique characteristics and intriguing molecular scaffolds. Here we report an enantioselective nickel-catalysed strategy for the construction of axially chiral alkenes via a 1,3-metallate shift of alkynyl tetracoordinate boron species. The chemoselectivity, regioselectivity and atroposelectivity can be regulated and well-controlled from readily accessible starting materials with a cheap transition-metal catalyst. Downstream transformations indicate the powerful conversion ability of such compounds in this protocol, and late-stage elaborations of bioactive compounds can also be achieved. Mechanistic experiments reveal that regioselective syn-addition of an aryl-Ni complex with a carbon-carbon triple bond and subsequent 1,3-phenyl migration are the two key steps for the synthesis of axially chiral alkenes.
Collapse
Affiliation(s)
- Xingxing Ma
- Key Laboratory of Molecule Synthesis and Function Discovery, Fujian Province University, College of Chemistry, Fuzhou University, Fuzhou, Fujian, China
| | - Mengwei Tan
- Key Laboratory of Molecule Synthesis and Function Discovery, Fujian Province University, College of Chemistry, Fuzhou University, Fuzhou, Fujian, China
| | - Luo Li
- Key Laboratory of Molecule Synthesis and Function Discovery, Fujian Province University, College of Chemistry, Fuzhou University, Fuzhou, Fujian, China
| | - Zihao Zhong
- Key Laboratory of Molecule Synthesis and Function Discovery, Fujian Province University, College of Chemistry, Fuzhou University, Fuzhou, Fujian, China
| | - Puhui Li
- Key Laboratory of Molecule Synthesis and Function Discovery, Fujian Province University, College of Chemistry, Fuzhou University, Fuzhou, Fujian, China
| | - Jinchao Liang
- Key Laboratory of Molecule Synthesis and Function Discovery, Fujian Province University, College of Chemistry, Fuzhou University, Fuzhou, Fujian, China
| | - Qiuling Song
- Key Laboratory of Molecule Synthesis and Function Discovery, Fujian Province University, College of Chemistry, Fuzhou University, Fuzhou, Fujian, China.
| |
Collapse
|
23
|
Krishna S, Echevarria KG, Reed CH, Eo H, Wintzinger M, Quattrocelli M, Valentine RJ, Selsby JT. A fat- and sucrose-enriched diet causes metabolic alterations in mdx mice. Am J Physiol Regul Integr Comp Physiol 2023; 325:R692-R711. [PMID: 37811713 PMCID: PMC11178302 DOI: 10.1152/ajpregu.00246.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 08/18/2023] [Accepted: 09/10/2023] [Indexed: 10/10/2023]
Abstract
Duchenne muscular dystrophy (DMD), a progressive muscle disease caused by the absence of functional dystrophin protein, is associated with multiple cellular, physiological, and metabolic dysfunctions. As an added complication to the primary insult, obesity/insulin resistance (O/IR) is frequently reported in patients with DMD; however, how IR impacts disease severity is unknown. We hypothesized a high-fat, high-sucrose diet (HFHSD) would induce O/IR, exacerbate disease severity, and cause metabolic alterations in dystrophic mice. To test this hypothesis, we treated 7-wk-old mdx (disease model) and C57 mice with a control diet (CD) or an HFHSD for 15 wk. The HFHSD induced insulin resistance, glucose intolerance, and hyperglycemia in C57 and mdx mice. Of note, mdx mice on CD were also insulin resistant. In addition, visceral adipose tissue weights were increased with HFHSD in C57 and mdx mice though differed by genotype. Serum creatine kinase activity and histopathological analyses using Masson's trichrome staining in the diaphragm indicated muscle damage was driven by dystrophin deficiency but was not augmented by diet. In addition, markers of inflammatory signaling, mitochondrial abundance, and autophagy were impacted by disease but not diet. Despite this, in addition to disease signatures in CD-fed mice, metabolomic and lipidomic analyses demonstrated a HFHSD caused some common changes in C57 and mdx mice and some unique signatures of O/IR within the context of dystrophin deficiency. In total, these data revealed that in mdx mice, 15 wk of HFHSD did not overtly exacerbate muscle injury but further impaired the metabolic status of dystrophic muscle.
Collapse
Affiliation(s)
- Swathy Krishna
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | | | - Carter H Reed
- Department of Kinesiology, Iowa State University, Ames, Iowa, United States
| | - Hyeyoon Eo
- Department of Kinesiology, Iowa State University, Ames, Iowa, United States
| | - Michelle Wintzinger
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Mattia Quattrocelli
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Rudy J Valentine
- Department of Kinesiology, Iowa State University, Ames, Iowa, United States
| | - Joshua T Selsby
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| |
Collapse
|
24
|
Garcia-Vallicrosa C, Falcon-Perez JM, Royo F. The Role of Longevity Assurance Homolog 2/Ceramide Synthase 2 in Bladder Cancer. Int J Mol Sci 2023; 24:15668. [PMID: 37958652 PMCID: PMC10650086 DOI: 10.3390/ijms242115668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
The human CERS2 gene encodes a ceramide synthase enzyme, known as CERS2 (ceramide synthase 2). This protein is also known as LASS2 (LAG1 longevity assurance homolog 2) and TMSG1 (tumor metastasis-suppressor gene 1). Although previously described as a tumor suppressor for different types of cancer, such as prostate or liver cancer, it has also been observed to promote tumor growth in adenocarcinoma. In this review, we focus on the influence of CERS2 in bladder cancer (BC), approaching the existing literature about its structure and activity, as well as the miRNAs regulating its expression. From a mechanistic point of view, different explanations for the role of CERS2 as an antitumor protein have been proposed, including the production of long-chain ceramides, interaction with vacuolar ATPase, and its function as inhibitor of mitochondrial fission. In addition, we reviewed the literature specifically studying the expression of this gene in both BC and biopsy-derived tumor cell lines, complementing this with an analysis of public gene expression data and its association with disease progression. We also discuss the importance of CERS2 as a biomarker and the presence of CERS2 mRNA in extracellular vesicles isolated from urine.
Collapse
Affiliation(s)
- Clara Garcia-Vallicrosa
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (C.G.-V.); (J.M.F.-P.)
| | - Juan M. Falcon-Perez
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (C.G.-V.); (J.M.F.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd), 28029 Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Felix Royo
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (C.G.-V.); (J.M.F.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas Y Digestivas (CIBERehd), 28029 Madrid, Spain
| |
Collapse
|
25
|
Ali-Berrada S, Guitton J, Tan-Chen S, Gyulkhandanyan A, Hajduch E, Le Stunff H. Circulating Sphingolipids and Glucose Homeostasis: An Update. Int J Mol Sci 2023; 24:12720. [PMID: 37628901 PMCID: PMC10454113 DOI: 10.3390/ijms241612720] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Sphingolipids are a family of lipid molecules produced through different pathways in mammals. Sphingolipids are structural components of membranes, but in response to obesity, they are implicated in the regulation of various cellular processes, including inflammation, apoptosis, cell proliferation, autophagy, and insulin resistance which favors dysregulation of glucose metabolism. Of all sphingolipids, two species, ceramides and sphingosine-1-phosphate (S1P), are also found abundantly secreted into the bloodstream and associated with lipoproteins or extracellular vesicles. Plasma concentrations of these sphingolipids can be altered upon metabolic disorders and could serve as predictive biomarkers of these diseases. Recent important advances suggest that circulating sphingolipids not only serve as biomarkers but could also serve as mediators in the dysregulation of glucose homeostasis. In this review, advances of molecular mechanisms involved in the regulation of ceramides and S1P association to lipoproteins or extracellular vesicles and how they could alter glucose metabolism are discussed.
Collapse
Affiliation(s)
- Sarah Ali-Berrada
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, F-75006 Paris, France; (S.A.-B.); (S.T.-C.); (A.G.)
- Institut Hospitalo-Universitaire ICAN, 75013 Paris, France
| | - Jeanne Guitton
- Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, CNRS UMR 9197, 91400 Saclay, France;
| | - Sophie Tan-Chen
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, F-75006 Paris, France; (S.A.-B.); (S.T.-C.); (A.G.)
- Institut Hospitalo-Universitaire ICAN, 75013 Paris, France
| | - Anna Gyulkhandanyan
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, F-75006 Paris, France; (S.A.-B.); (S.T.-C.); (A.G.)
- Institut Hospitalo-Universitaire ICAN, 75013 Paris, France
| | - Eric Hajduch
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, F-75006 Paris, France; (S.A.-B.); (S.T.-C.); (A.G.)
- Institut Hospitalo-Universitaire ICAN, 75013 Paris, France
| | - Hervé Le Stunff
- Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, CNRS UMR 9197, 91400 Saclay, France;
| |
Collapse
|
26
|
Watt NT, McGrane A, Roberts LD. Linking the unfolded protein response to bioactive lipid metabolism and signalling in the cell non-autonomous extracellular communication of ER stress. Bioessays 2023; 45:e2300029. [PMID: 37183938 PMCID: PMC11475223 DOI: 10.1002/bies.202300029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/16/2023]
Abstract
The endoplasmic reticulum (ER) organelle is the key intracellular site of both protein and lipid biosynthesis. ER dysfunction, termed ER stress, can result in protein accretion within the ER and cell death; a pathophysiological process contributing to a range of metabolic diseases and cancers. ER stress leads to the activation of a protective signalling cascade termed the Unfolded Protein Response (UPR). However, chronic UPR activation can ultimately result in cellular apoptosis. Emerging evidence suggests that cells undergoing ER stress and UPR activation can release extracellular signals that can propagate UPR activation to target tissues in a cell non-autonomous signalling mechanism. Separately, studies have determined that the UPR plays a key regulatory role in the biosynthesis of bioactive signalling lipids including sphingolipids and ceramides. Here we weigh the evidence to combine these concepts and propose that during ER stress, UPR activation drives the biosynthesis of ceramide lipids, which are exported and function as cell non-autonomous signals to propagate UPR activation in target cells and tissues.
Collapse
Affiliation(s)
- Nicole T. Watt
- Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Anna McGrane
- Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Lee D. Roberts
- Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| |
Collapse
|
27
|
Tzou FY, Hornemann T, Yeh JY, Huang SY. The pathophysiological role of dihydroceramide desaturase in the nervous system. Prog Lipid Res 2023; 91:101236. [PMID: 37187315 DOI: 10.1016/j.plipres.2023.101236] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 04/18/2023] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
Dihydroceramide desaturase 1 (DEGS1) converts dihydroceramide (dhCer) to ceramide (Cer) by inserting a C4-C5 trans (∆4E) double bond into the sphingoid backbone. Low DEGS activity causes accumulation of dhCer and other dihydrosphingolipid species. Although dhCer and Cer are structurally very similar, their imbalances can have major consequences both in vitro and in vivo. Mutations in the human DEGS1 gene are known to cause severe neurological defects, such as hypomyelinating leukodystrophy. Likewise, inhibition of DEGS1 activity in fly and zebrafish models causes dhCer accumulation and subsequent neuronal dysfunction, suggesting that DEGS1 activity plays a conserved and critical role in the nervous system. Dihydrosphingolipids and their desaturated counterparts are known to control various essential processes, including autophagy, exosome biogenesis, ER stress, cell proliferation, and cell death. Furthermore, model membranes with either dihydrosphingolipids or sphingolipids exhibit different biophysical properties, including membrane permeability and packing, thermal stability, and lipid diffusion. However, the links between molecular properties, in vivo functional data, and clinical manifestations that underlie impaired DEGS1 function remain largely unresolved. In this review, we summarize the known biological and pathophysiological roles of dhCer and its derivative dihydrosphingolipid species in the nervous system, and we highlight several possible disease mechanisms that warrant further investigation.
Collapse
Affiliation(s)
- Fei-Yang Tzou
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Thorsten Hornemann
- Institute for Clinical Chemistry, University Hospital and University Zurich, 8091 Zürich, Switzerland
| | - Jui-Yu Yeh
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Yi Huang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
28
|
Smith S, Lopez S, Kim A, Kasteri J, Olumuyide E, Punu K, de la Parra C, Sauane M. Interleukin 24: Signal Transduction Pathways. Cancers (Basel) 2023; 15:3365. [PMID: 37444474 PMCID: PMC10340555 DOI: 10.3390/cancers15133365] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/16/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Interleukin 24 is a member of the IL-10 family with crucial roles in antitumor, wound healing responses, host defense, immune regulation, and inflammation. Interleukin 24 is produced by both immune and nonimmune cells. Its canonical pathway relies on recognition and interaction with specific Interleukin 20 receptors in the plasma membrane and subsequent cytoplasmic Janus protein tyrosine kinases (JAK)/signal transducer and activator of the transcription (STAT) activation. The identification of noncanonical JAK/STAT-independent signaling pathways downstream of IL-24 relies on the interaction of IL-24 with protein kinase R in the cytosol, respiratory chain proteins in the inner mitochondrial membrane, and chaperones such as Sigma 1 Receptor in the endoplasmic reticulum. Numerous studies have shown that enhancing or inhibiting the expression of Interleukin 24 has a therapeutic effect in animal models and clinical trials in different pathologies. Successful drug targeting will require a deeper understanding of the downstream signaling pathways. In this review, we discuss the signaling pathway triggered by IL-24.
Collapse
Affiliation(s)
- Simira Smith
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; (S.S.); (S.L.); (J.K.); (E.O.); (K.P.)
| | - Sual Lopez
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; (S.S.); (S.L.); (J.K.); (E.O.); (K.P.)
| | - Anastassiya Kim
- Ph.D. Program in Biology, The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA; (A.K.); (C.d.l.P.)
| | - Justina Kasteri
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; (S.S.); (S.L.); (J.K.); (E.O.); (K.P.)
| | - Ezekiel Olumuyide
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; (S.S.); (S.L.); (J.K.); (E.O.); (K.P.)
| | - Kristian Punu
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; (S.S.); (S.L.); (J.K.); (E.O.); (K.P.)
| | - Columba de la Parra
- Ph.D. Program in Biology, The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA; (A.K.); (C.d.l.P.)
- Department of Chemistry, Herbert H. Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA
| | - Moira Sauane
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, 250 Bedford Park Boulevard West, Bronx, NY 10468, USA; (S.S.); (S.L.); (J.K.); (E.O.); (K.P.)
- Ph.D. Program in Biology, The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA; (A.K.); (C.d.l.P.)
| |
Collapse
|
29
|
Cao R, Tian H, Zhang Y, Liu G, Xu H, Rao G, Tian Y, Fu X. Signaling pathways and intervention for therapy of type 2 diabetes mellitus. MedComm (Beijing) 2023; 4:e283. [PMID: 37303813 PMCID: PMC10248034 DOI: 10.1002/mco2.283] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/18/2023] [Accepted: 04/27/2023] [Indexed: 06/13/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) represents one of the fastest growing epidemic metabolic disorders worldwide and is a strong contributor for a broad range of comorbidities, including vascular, visual, neurological, kidney, and liver diseases. Moreover, recent data suggest a mutual interplay between T2DM and Corona Virus Disease 2019 (COVID-19). T2DM is characterized by insulin resistance (IR) and pancreatic β cell dysfunction. Pioneering discoveries throughout the past few decades have established notable links between signaling pathways and T2DM pathogenesis and therapy. Importantly, a number of signaling pathways substantially control the advancement of core pathological changes in T2DM, including IR and β cell dysfunction, as well as additional pathogenic disturbances. Accordingly, an improved understanding of these signaling pathways sheds light on tractable targets and strategies for developing and repurposing critical therapies to treat T2DM and its complications. In this review, we provide a brief overview of the history of T2DM and signaling pathways, and offer a systematic update on the role and mechanism of key signaling pathways underlying the onset, development, and progression of T2DM. In this content, we also summarize current therapeutic drugs/agents associated with signaling pathways for the treatment of T2DM and its complications, and discuss some implications and directions to the future of this field.
Collapse
Affiliation(s)
- Rong Cao
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Huimin Tian
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yu Zhang
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Geng Liu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Haixia Xu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Guocheng Rao
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yan Tian
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Xianghui Fu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
30
|
Meadows AM, Han K, Singh K, Murgia A, McNally BD, West JA, Huffstutler RD, Powell-Wiley TM, Baumer Y, Griffin JL, Sack MN. N-arachidonylglycine is a caloric state-dependent circulating metabolite which regulates human CD4 +T cell responsiveness. iScience 2023; 26:106578. [PMID: 37128607 PMCID: PMC10148119 DOI: 10.1016/j.isci.2023.106578] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/29/2023] [Accepted: 03/29/2023] [Indexed: 05/03/2023] Open
Abstract
Caloric deprivation interventions such as intermittent fasting and caloric restriction ameliorate metabolic and inflammatory disease. As a human model of caloric deprivation, a 24-h fast blunts innate and adaptive immune cell responsiveness relative to the refed state. Isolated serum at these time points confers these same immunomodulatory effects on transformed cell lines. To identify serum mediators orchestrating this, metabolomic and lipidomic analysis was performed on serum extracted after a 24-h fast and re-feeding. Bioinformatic integration with concurrent peripheral blood mononuclear cells RNA-seq analysis implicated key metabolite-sensing GPCRs in fasting-mediated immunomodulation. The putative GPR18 ligand N-arachidonylglycine (NAGly) was elevated during fasting and attenuated CD4+T cell responsiveness via GPR18 MTORC1 signaling. In parallel, NAGly reduced inflammatory Th1 and Th17 cytokines levels in CD4+T cells isolated from obese subjects, identifying a fasting-responsive metabolic intermediate that may contribute to the regulation of nutrient-level dependent inflammation associated with metabolic disease.
Collapse
Affiliation(s)
- Allison M. Meadows
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, MD, USA
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Kim Han
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, MD, USA
| | - Komudi Singh
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, MD, USA
| | - Antonio Murgia
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Ben D. McNally
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - James A. West
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | | | - Tiffany M. Powell-Wiley
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, NHLBI, NIH, Bethesda, MD, USA
| | - Yvonne Baumer
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, NHLBI, NIH, Bethesda, MD, USA
| | - Julian L. Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
- The Rowett Institute, School of Medicine, Medical Sciences and Nutrition, Foresterhill Campus, Aberdeen, UK
| | - Michael N. Sack
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, MD, USA
- Cardiovascular Branch, NHLBI, NIH, Bethesda, MD, USA
- Corresponding author
| |
Collapse
|
31
|
Raines LN, Huang SCC. How the Unfolded Protein Response Is a Boon for Tumors and a Bane for the Immune System. Immunohorizons 2023; 7:256-264. [PMID: 37067519 PMCID: PMC10579845 DOI: 10.4049/immunohorizons.2200064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/28/2023] [Indexed: 04/18/2023] Open
Abstract
The correct folding of proteins is essential for appropriate cell function and is tightly regulated within the endoplasmic reticulum (ER). Environmental challenges and cellular conditions disrupt ER homeostasis and induce ER stress, which adversely affect protein folding and activate the unfolded protein response (UPR). It is now becoming recognized that cancer cells can overcome survival challenges posed within the tumor microenvironment by activating the UPR. Furthermore, the UPR has also been found to impose detrimental effects on immune cells by inducing immunoinhibitory activity in both tumor-infiltrating innate and adaptive immune cells. This suggests that these signaling axes may be important therapeutic targets, resulting in multifaceted approaches to eradicating tumor cells. In this mini-review, we discuss the role of the UPR in driving tumor progression and modulating the immune system's ability to target cancer cells. Additionally, we highlight some of the key unanswered questions that may steer future UPR research.
Collapse
Affiliation(s)
- Lydia N. Raines
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Stanley Ching-Cheng Huang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH
| |
Collapse
|
32
|
Abstract
During aging, animals experience a decline in proteostasis activity, including loss of stress-response activation, culminating in the accumulation of misfolded proteins and toxic aggregates, which are causal in the onset of some chronic diseases. Finding genetic and pharmaceutical treatments that can increase organismal proteostasis and lengthen life is an ongoing goal of current research. The regulation of stress responses by cell non-autonomous mechanisms appears to be a potent way to impact organismal healthspan. In this Review, we cover recent findings in the intersection of proteostasis and aging, with a special focus on articles and preprints published between November 2021 and October 2022. A significant number of papers published during this time increased our understanding of how cells communicate with each other during proteotoxic stress. Finally, we also draw attention to emerging datasets that can be explored to generate new hypotheses that explain age-related proteostasis collapse.
Collapse
Affiliation(s)
- Maximilian A. Thompson
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Evandro A. De-Souza
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| |
Collapse
|
33
|
Zhu C, Huai Q, Zhang X, Dai H, Li X, Wang H. Insights into the roles and pathomechanisms of ceramide and sphigosine-1-phosphate in nonalcoholic fatty liver disease. Int J Biol Sci 2023; 19:311-330. [PMID: 36594091 PMCID: PMC9760443 DOI: 10.7150/ijbs.78525] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/12/2022] [Indexed: 11/24/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), as one of the main causes of chronic liver disease worldwide, encompasses a spectrum of liver conditions that are not caused by other etiology, such as overt alcohol consumption, from simple steatosis to more aggressive non-alcoholic steatohepatitis (NASH) that involves liver inflammation and fibrosis, and to the lethal cirrhosis that may result in liver cancer and liver failure. The molecular mechanisms governing the transition from steatosis to NASH remain not fully understood, but the hepatic lipidome is extensively altered in the setting of steatosis and steatohepatitis, which also correlate with disease progression. With the tremendous advancement in the field of lipidomics in last two decades, a better understanding of the specific role of sphingolipids in fatty liver disease has taken shape. Among the numerous lipid subtypes that accumulate, ceramides are particularly impactful. On the one hand, excessive ceramides deposition in the liver cause hepatic steatosis. On the other hand, ceramides as lipotoxic lipid have significant effects on hepatic inflammation, apoptosis and insulin resistance that contribute to NAFLD. In this review, we summarize and evaluate current understanding of the multiple roles of ceramides in the onset of fatty liver disease and the pathogenic mechanisms underlying their effects, and we also discuss recent advances and challenges in pharmacological interventions targeting ceramide metabolism for the treatment of NAFLD.
Collapse
Affiliation(s)
- Cheng Zhu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qian Huai
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xu Zhang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hanren Dai
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiaolei Li
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
34
|
Yang Z, Huo Y, Zhou S, Guo J, Ma X, Li T, Fan C, Wang L. Cancer cell-intrinsic XBP1 drives immunosuppressive reprogramming of intratumoral myeloid cells by promoting cholesterol production. Cell Metab 2022; 34:2018-2035.e8. [PMID: 36351432 DOI: 10.1016/j.cmet.2022.10.010] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 08/24/2022] [Accepted: 10/14/2022] [Indexed: 11/09/2022]
Abstract
A hostile microenvironment in tumor tissues disrupts endoplasmic reticulum homeostasis and induces the unfolded protein response (UPR). A chronic UPR in both cancer cells and tumor-infiltrating leukocytes could facilitate the evasion of immune surveillance. However, how the UPR in cancer cells cripples the anti-tumor immune response is unclear. Here, we demonstrate that, in cancer cells, the UPR component X-box binding protein 1 (XBP1) favors the synthesis and secretion of cholesterol, which activates myeloid-derived suppressor cells (MDSCs) and causes immunosuppression. Cholesterol is delivered in the form of small extracellular vesicles and internalized by MDSCs through macropinocytosis. Genetic or pharmacological depletion of XBP1 or reducing the tumor cholesterol content remarkably decreases MDSC abundance and triggers robust anti-tumor responses. Thus, our data unravel the cell-non-autonomous role of XBP1/cholesterol signaling in the regulation of tumor growth and suggest its inhibition as a useful strategy for improving the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Zaili Yang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yazhen Huo
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Shixin Zhou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingya Guo
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaotu Ma
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Tao Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Congli Fan
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Likun Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
35
|
Kundu P, Paraiso IL, Choi J, Miranda CL, Kioussi C, Maier CS, Bobe G, Stevens JF, Raber J. Xanthohumol improves cognition in farnesoid X receptor-deficient mice on a high-fat diet. Dis Model Mech 2022; 15:dmm049820. [PMID: 36353888 PMCID: PMC9713832 DOI: 10.1242/dmm.049820] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/26/2022] [Indexed: 08/18/2023] Open
Abstract
Xanthohumol (XN) improves cognition of wild-type rodents on a high-fat diet (HFD). Bile acids and ceramide levels in the liver and hippocampus might be linked to these effects. XN modulates activity of the nuclear farnesoid X receptor (FXR; also known as NR1H4), the primary receptor for bile acids. To determine the role of FXR in the liver and intestine in mediating the effects of XN on cognitive performance, mice with intestine- and liver-specific FXR ablation (FXRIntestine-/- and FXRLiver-/-, respectively) on an HFD or an HFD containing XN were cognitively tested. XN improved cognitive performance in a genotype- and sex-dependent manner, with improved task learning in females (specifically wild-type), reversal learning in males (specifically wild-type and FXRIntestine-/- mutant) and spatial learning (both sexes). XN increased hippocampal diacylglycerol and sphingomyelin levels in females but decreased them in males. XN increased the ratio of shorter-chain to longer-chain ceramides and hexaceramides. Higher diacylglycerol and lower longer-chain ceramide and hexaceramide levels were linked to improved cognitive performance. Thus, the beneficial sex-dependent cognitive effects of XN are linked to changes in hippocampal diacylglycerol and ceramide levels. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Payel Kundu
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239, USA
| | - Ines L. Paraiso
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Jaewoo Choi
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Cristobal L. Miranda
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Claudia S. Maier
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA
| | - Gerd Bobe
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Jan F. Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239, USA
- Departments of Neurology and Radiation Medicine, Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
36
|
Cabezas Perez RJ, Ávila Rodríguez MF, Rosero Salazar DH. Exogenous Antioxidants in Remyelination and Skeletal Muscle Recovery. Biomedicines 2022; 10:biomedicines10102557. [PMID: 36289819 PMCID: PMC9599955 DOI: 10.3390/biomedicines10102557] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Inflammatory, oxidative, and autoimmune responses cause severe damage to the nervous system inducing loss of myelin layers or demyelination. Even though demyelination is not considered a direct cause of skeletal muscle disease there is extensive damage in skeletal muscles following demyelination and impaired innervation. In vitro and in vivo evidence using exogenous antioxidants in models of demyelination is showing improvements in myelin formation alongside skeletal muscle recovery. For instance, exogenous antioxidants such as EGCG stimulate nerve structure maintenance, activation of glial cells, and reduction of oxidative stress. Consequently, this evidence is also showing structural and functional recovery of impaired skeletal muscles due to demyelination. Exogenous antioxidants mostly target inflammatory pathways and stimulate remyelinating mechanisms that seem to induce skeletal muscle regeneration. Therefore, the aim of this review is to describe recent evidence related to the molecular mechanisms in nerve and skeletal muscle regeneration induced by exogenous antioxidants. This will be relevant to identifying further targets to improve treatments of neuromuscular demyelinating diseases.
Collapse
|
37
|
Camacho-Muñoz D, Niven J, Kucuk S, Cucchi D, Certo M, Jones SW, Fischer DP, Mauro C, Nicolaou A. Omega-3 polyunsaturated fatty acids reverse the impact of western diets on regulatory T cell responses through averting ceramide-mediated pathways. Biochem Pharmacol 2022; 204:115211. [PMID: 35985403 DOI: 10.1016/j.bcp.2022.115211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 11/02/2022]
Abstract
Western diet (WD), high in sugar and fat, promotes obesity and associated chronic low-grade pro-inflammatory environment, leading to impaired immune function, reprogramming of innate and adaptive immune cells, and development of chronic degenerative diseases, including cardiovascular disease. Increased concentrations of circulating and tissue ceramides contribute to inflammation and cellular dysfunction common in immune metabolic and cardiometabolic disease. Therefore, ceramide-lowering interventions have been considered as strategies to improve adipose tissue health. Here, we report the ability of omega-3 polyunsaturated fatty acids (n-3PUFA) to attenuate inflammatory phenotypes promoted by WD, through ceramide-dependent pathways. Using an animal model, we show that enrichment of WD diet with n-3PUFA, reduced the expression of ceramide synthase 2 (CerS2), and lowered the concentration of long-chain ceramides (C23-C26) in plasma and adipose tissues. N-3PUFA also increased prevalence of the anti-inflammatory CD4+Foxp3+ and CD4+Foxp3+CD25+ Treg subtypes in lymphoid organs. The CerS inhibitor FTY720 mirrored the effect of n-3PUFA. Treatment of animal and human T cells with ceramide C24 in vitro, reduced CD4+Foxp3+ Treg polarisation and IL-10 production, and increased IL-17, while it decreased Erk and Akt phosphorylation downstream of T cell antigen receptors (TCR). These findings suggest that molecular mechanisms mediating the adverse effect of ceramides on regulatory T lymphocytes, progress through reduced TCR signalling. Our findings suggest that nutritional enrichment of WD with fish oil n-3PUFA can partially mitigate its detrimental effects, potentially improving the low-grade inflammation associated with immune metabolic disease. Compared to pharmacological interventions, n-3PUFA offer a simpler approach that can be accommodated as lifestyle choice.
Collapse
Affiliation(s)
- Dolores Camacho-Muñoz
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK
| | - Jennifer Niven
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2WB, UK
| | - Salih Kucuk
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2WB, UK
| | - Danilo Cucchi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Michelangelo Certo
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2WB, UK
| | - Simon W Jones
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2WB, UK
| | - Deborah P Fischer
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK
| | - Claudio Mauro
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2WB, UK; William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Anna Nicolaou
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, UK.
| |
Collapse
|
38
|
Höring M, Peschel G, Grimm J, Krautbauer S, Müller M, Weigand K, Liebisch G, Buechler C. Serum Ceramide Species Are Associated with Liver Cirrhosis and Viral Genotype in Patients with Hepatitis C Infection. Int J Mol Sci 2022; 23:ijms23179806. [PMID: 36077197 PMCID: PMC9456360 DOI: 10.3390/ijms23179806] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/22/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Hepatitis C virus (HCV) infection affects ceramide metabolism, and, here, we have evaluated associations of eight serum ceramide species with viral load, viral genotype, and disease markers in 178 patients with chronic HCV. In this cohort, ceramide d18:1;O2/16:0 was higher in the serum of the 20 diabetic patients compared to the patients without this complication. Moreover, ceramide d18:1;O2/24:0 was negatively correlated with age. Of note, all but ceramide d18:1;O2/16:0 and 26:0 were diminished in the serum of patients with liver cirrhosis and, with the exception of ceramide d18:1;O2/16:0, were negatively correlated with the model for end-stage liver disease (MELD) score. Most of the serum ceramides are carried in low-density lipoprotein (LDL), which rises following effective direct-acting antiviral (DAA) therapy. Ceramide d18:1;O2/24:0 recovered in parallel with LDL, whereas ceramide d18:1;O2/18:0 declined. Genotype-3-infected patients had the lowest ceramide levels, which were comparable to other genotypes after DAA treatment. Notably, ceramide d18:1;O2/23:0 and 24:0 were negatively correlated with the MELD score in patients with liver cirrhosis at the end of DAA therapy. Long-chain (LC) ceramides show adverse effects, whereas very-long-chain (VL) species have protective functions in the liver. The ratio of VL/LC ceramides was higher in non-cirrhosis patients than cirrhosis patients and further increased at the end of therapy in this subgroup. In summary, our study shows that serum ceramide levels are related to liver cirrhosis and viral genotype. Whether the more favorable serum ceramide profile in non-cirrhosis patients, before and after DAA therapy, is of pathophysiological importance needs further investigation.
Collapse
Affiliation(s)
- Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Georg Peschel
- Department of Internal Medicine I, University Hospital Regensburg, 93053 Regensburg, Germany
- Department of Internal Medicine, Klinikum Fürstenfeldbruck, 82256 Fürstenfeldbruck, Germany
| | - Jonathan Grimm
- Department of Internal Medicine I, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Sabrina Krautbauer
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Martina Müller
- Department of Internal Medicine I, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Kilian Weigand
- Department of Internal Medicine I, University Hospital Regensburg, 93053 Regensburg, Germany
- Department of Gastroenterology, Gemeinschaftsklinikum Mittelrhein, 56073 Koblenz, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Christa Buechler
- Department of Internal Medicine I, University Hospital Regensburg, 93053 Regensburg, Germany
- Correspondence: ; Tel.: +49-941-944-7009
| |
Collapse
|